Antonio Jesús Hinojosa, Rubén Deogracias, Beatriz Rico  Cell Reports 

Slides:



Advertisements
Similar presentations
Volume 22, Issue 16, Pages (August 2012)
Advertisements

Volume 19, Issue 8, Pages (May 2017)
Johanna Sigl-Glöckner, Michael Brecht  Cell Reports 
Volume 82, Issue 1, Pages (April 2014)
Volume 49, Issue 6, Pages (March 2006)
Volume 21, Issue 9, Pages (November 2017)
Volume 71, Issue 5, Pages (September 2011)
History-Dependent Catastrophes Regulate Axonal Microtubule Behavior
Volume 94, Issue 2, Pages e7 (April 2017)
Volume 8, Issue 4, Pages (April 2017)
Golgi Outposts Shape Dendrite Morphology by Functioning as Sites of Acentrosomal Microtubule Nucleation in Neurons  Kassandra M. Ori-McKenney, Lily Yeh.
Volume 24, Issue 22, Pages (November 2014)
Volume 24, Issue 1, Pages e9 (July 2018)
Volume 23, Issue 7, Pages (May 2018)
Hai-Yan He, Wanhua Shen, Masaki Hiramoto, Hollis T. Cline  Neuron 
Dante S. Bortone, Shawn R. Olsen, Massimo Scanziani  Neuron 
Euiseok J. Kim, Matthew W. Jacobs, Tony Ito-Cole, Edward M. Callaway 
Volume 96, Issue 4, Pages e5 (November 2017)
Odor Processing by Adult-Born Neurons
Volume 11, Issue 12, Pages (June 2015)
Volume 89, Issue 5, Pages (March 2016)
Volume 19, Issue 8, Pages (May 2017)
Volume 13, Issue 9, Pages (December 2015)
Volume 13, Issue 2, Pages (January 2003)
High Plasticity of New Granule Cells in the Aging Hippocampus
Johanna Sigl-Glöckner, Michael Brecht  Cell Reports 
Transient and Persistent Dendritic Spines in the Neocortex In Vivo
Volume 91, Issue 2, Pages (July 2016)
EB3 Regulates Microtubule Dynamics at the Cell Cortex and Is Required for Myoblast Elongation and Fusion  Anne Straube, Andreas Merdes  Current Biology 
Functional Distinctions between Spine and Dendritic Synapses Made onto Parvalbumin- Positive Interneurons in Mouse Cortex  Laura Sancho, Brenda L. Bloodgood 
Γ-Protocadherins Control Cortical Dendrite Arborization by Regulating the Activity of a FAK/PKC/MARCKS Signaling Pathway  Andrew M. Garrett, Dietmar Schreiner,
Volume 21, Issue 3, Pages (October 2017)
Volume 17, Issue 9, Pages (November 2016)
Volume 21, Issue 13, Pages (December 2017)
Volume 10, Issue 4, Pages (April 2018)
Volume 89, Issue 3, Pages (February 2016)
Elizabeth A.K. Phillips, Christoph E. Schreiner, Andrea R. Hasenstaub 
Volume 18, Issue 11, Pages (March 2017)
Experience-Dependent Equilibration of AMPAR-Mediated Synaptic Transmission during the Critical Period  Kyung-Seok Han, Samuel F. Cooke, Weifeng Xu  Cell.
Volume 20, Issue 13, Pages (September 2017)
Volume 54, Issue 6, Pages (June 2007)
Volume 20, Issue 6, Pages (August 2017)
Rui Qin, Shuai Cao, Tianjie Lyu, Cai Qi, Weiguang Zhang, Yun Wang 
Matthew S. Kayser, Mark J. Nolt, Matthew B. Dalva  Neuron 
Volume 22, Issue 7, Pages (February 2018)
Ashkan Javaherian, Hollis T. Cline  Neuron 
Han Xu, Hyo-Young Jeong, Robin Tremblay, Bernardo Rudy  Neuron 
Volume 12, Issue 11, Pages (September 2015)
Volume 65, Issue 3, Pages (February 2010)
Xuepei Lei, Jianwei Jiao  Stem Cell Reports 
Giulia Quattrocolo, Gord Fishell, Timothy J. Petros  Cell Reports 
Target-Specific Glycinergic Transmission from VGluT3-Expressing Amacrine Cells Shapes Suppressive Contrast Responses in the Retina  Nai-Wen Tien, Tahnbee.
Volume 12, Issue 10, Pages (September 2015)
Volume 21, Issue 3, Pages (October 2017)
Volume 26, Issue 19, Pages (October 2016)
Volume 27, Issue 5, Pages e6 (April 2019)
Volume 17, Issue 7, Pages (November 2016)
Volume 92, Issue 6, Pages (December 2016)
Corticostriatal Transmission Is Selectively Enhanced in Striatonigral Neurons with Postnatal Loss of Tsc1  Katelyn N. Benthall, Stacie L. Ong, Helen S.
Volume 13, Issue 12, Pages (December 2015)
Three-Step Model for Polarized Sorting of KIF17 into Dendrites
Spontaneous Neurotransmitter Release Shapes Dendritic Arbors via Long-Range Activation of NMDA Receptors  Laura C. Andreae, Juan Burrone  Cell Reports 
Volume 17, Issue 2, Pages (October 2016)
Volume 27, Issue 7, Pages (April 2017)
Volume 18, Issue 1, Pages (January 2017)
Giulia Quattrocolo, Gord Fishell, Timothy J. Petros  Cell Reports 
Volume 26, Issue 8, Pages e6 (February 2019)
Irreplaceability of Neuronal Ensembles after Memory Allocation
Volume 8, Issue 3, Pages (March 2017)
Feedback-Driven Assembly of the Axon Initial Segment
Presentation transcript:

The Microtubule Regulator NEK7 Coordinates the Wiring of Cortical Parvalbumin Interneurons  Antonio Jesús Hinojosa, Rubén Deogracias, Beatriz Rico  Cell Reports  Volume 24, Issue 5, Pages 1231-1242 (July 2018) DOI: 10.1016/j.celrep.2018.06.115 Copyright © 2018 Terms and Conditions

Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions

Figure 1 Nek7 Transcripts Are Upregulated during GABAergic Interneuron Wiring and Expressed Mostly in PV+ Cells (A) Schematic of genetic screening. Cortices from different GFP+ reporter lines were dissected, and cells were dissociated and FACS sorted and their RNA hybridized to microarrays. (B) Bioinformatic comparisons of neuronal populations. (C) Heatmap showing the expression levels of the 20 first ranked genes. Nek7 is highlighted in red. (D) Confocal images showing in situ hybridization for Nek7 (red) and immunohistochemistry for GFP (green) and SST+ (cyan) in the somatosensory cortex of P10 Lhx6Cre;RCE mice. GFP+ and SST+ are somatostatin cells, GFP+ and SST− are putative PV+ interneurons. (E) Confocal images showing in situ hybridization for Nek7 (red) and immunohistochemistry for PV+ cells (green) in the somatosensory cortex of P30 wild-type mice. In (D) and (E), filled arrowheads denote colocalization, and open arrowheads denote no colocalization. (F) Percentage of PV+ (gray) among all Nek7-expressing cortical cells at P10 and P30. (G) Percentage of Nek7-positive cells among all PV-expressing neurons at P10 and P30. IN, interneurons (red); Pyr, pyramidal cells (blue). Data are represented as mean ± SEM. Scale bar represents 50 μm. See also Figures S1–S4. Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions

Figure 2 Nek7 Knockdown Accelerates Microtubule Growth (A) Diagram of the Cre-dependent constructs expressing mCherry and shRNA. The plasmids were co-transfected with EB3-YFP, and mNek7 for the rescue, in primary cortical cultures from Nkx2-1Cre mice at 4 DIV, and axons were recorded at 7 DIV. (B–D) Confocal Z projection frames from control shRNA (B), Nek7 shRNA (C), and rescue with mNek7 (D) Nkx2-1Cre growth cones expressing mCherry (red, before the time-lapse) and EB3-YFP (gray, time-lapse). The path of EB3-YFP comets is tracked with a red line. Scale bar represents 2 μm. (E–G) Kymographs of EB3 comets in control (E), Nek7 shRNA (F), and rescue (G) growth cones showing their existence time as a function of distance. (H and I) Average speed (H) and speed distribution of EB3 comets (I) comparing control (n = 31 growth cones), Nek7-depleted cells (n = 35 growth cones), and mNek7 rescue cells (n = 35 growth cones) from three independent cultures. All growth cones with EB3 comets were quantified. One-way ANOVA (H) and χ2 test (I). ∗p < 0.05 and ∗∗p < 0.01. Data are represented as mean ± SEM (H) or total cell percentage (I). See also Figures S5–S7 and Videos S1, S2, and S3. Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions

Figure 3 Nek7 Depletion Impairs Axonal Growth Cone Dynamics In Vitro (A) Schematic of experimental design with the Cre-dependent plasmids transfected in Nkx2-1Cre primary cortical cultures at 4 DIV and axons recorded at 7 DIV. (B–D) Confocal Z projection frames from axons expressing control (B), Nek7 shRNA (C), and mNek7 rescue (D). The last frame in the sequence shows superimposed images of the frames t = 0 min (red), t = 15 min (green), t = 30 min (blue), and t = 45 min (white). Scale bar represents 5 μm. (E–G) Axonal speed (E), average of growth cone maximum turning angle (F), and its distribution (G) from cells expressing control shRNA (n = 64 growth cones), Nek7 shRNA (n = 59 growth cones), and mNek7 (n = 85 growth cones) from three or four independent cultures. All growth cones increasing in length were quantified. Kruskal-Wallis test, pairwise comparisons (E and F), and χ2 test (G). Data are represented as mean ± SEM (E and F) or total cell percentage (G). ∗∗p < 0.01 and ∗∗∗p < 0.001; n.s., not significant. See also Figures S6 and S7 and Videos S4, S5, and S6. Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions

Figure 4 Loss of NEK7 Alters Interneuron Morphology In Vitro (A) Schematic of experimental design. pDIO-shRNA was transfected in Nkx2-1Cre primary cortical cultures. (B–D) Confocal Z projections of Nkx2-1Cre interneurons from cortical cultures transfected with control shRNA (B), Nek7 shRNA (C), and mNek7 for rescue (D) expressing mCherry. The cells were automatically reconstructed and masked in black at 12 DIV. Scale bars represent 200 μm. (E–I) Average of total neurite length (E) and its distribution (F), Sholl analysis (G), total branching points (H), and branching points per unit length (I) of control shRNA (n = 47), Nek7 shRNA (n = 45), and rescue (n = 46) transfected neurons from three or four independent cultures. All imaged cells were quantified. Kruskal-Wallis test, pairwise comparisons (E, H, and I), χ2 test (F), and two-way ANOVA with Bonferroni correction (G). ∗p < 0.05, ∗∗p < 0.01, and ∗∗∗p < 0.001. Data are represented as mean ± SEM. See also Figures S6 and S7. Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions

Figure 5 Nek7 Overexpression in Pyramids Does Not Alter Their Axonal Development (A and B) Confocal Z projection frames from NexCre growth cones expressing mCherry (red, before the time-lapse), EB3-YFP (gray, time-lapse), and mNek7 in the Nek7 condition. Control (A) and mNek7 (B). The path of EB3-YFP comets is tracked with a red line. (C and D) Confocal Z projection frames from NexCre growth cones. Control (C) and mNEK7 (D). The last frame in the sequence shows superimposed images of the frames t = 0 min (red), t = 15 min (green), t = 30 min (blue), and t = 45 min (white). (E and F) Confocal Z projections of NexCre interneurons expressing mCherry. Control (E) and mNek7 (F). The cells were automatically reconstructed and masked in black at 8 DIV. (G) Average speed of EB3 comets comparing control pyramids (A; n = 53 growth cones) and Nek7-expressing pyramids (B; n = 54 growth cones) from three independent cultures. (H and I) Average of growth cone maximum turning angle (H) and axonal speed (I) from control pyramids (C; n = 73 growth cones) and Nek7-expressing pyramids (D; n = 74 growth cones) from three independent cultures. (J and K) Neurite length (J) and branching points (K) comparing control pyramids (E; n = 44 cells) and Nek7-expressing pyramids (F; n = 38 cells) transfected neurons from three independent cultures. One-way ANOVA (G and I) and Mann-Whitney test (H, J, and K). n.s., not significant. Data are represented as mean ± SEM. Scale bars represent 2 μm (A and B), 5 μm (C and D), and 200 μm (E and F). Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions

Figure 6 Loss of NEK7 Causes Abnormal PV+ Neuronal Morphology In Vivo (A) Schematic of experimental design. Cre-dependent AAVs expressing shRNA and the fluorescent marker mCherry were injected in E15.5 Lhx6Cre mice in vivo. (B and C) Targeted mCherry+ interneurons (B) that were PV+ (C) were selected for the analysis. (D and E) Confocal Z projections of PV+ interneurons expressing control shRNA (D) and Nek7 shRNA (E) reported by mCherry. The cells were automatically reconstructed and masked in black from layer V of somatosensory cortex at P21. (F–I) Average of total neurite length (F), Sholl analysis (G), total branching points (H), and branching points per 100 μm of neurite (I) of control shRNA (n = 30 PV+ neurons, from 10 mice) and Nek7 shRNA (n = 27 PV+ neurons, from 8 mice) infected neurons. One-way ANOVA (F, H, and I) and two-way ANOVA with Bonferroni correction (G). ∗p < 0.05, ∗∗p < 0.01, and ∗∗∗p < 0.001; n.s., not significant. Data are represented as mean ± SEM. Scale bars represent 200 μm (B) and 50 μm (C–E). See also Figure S6. Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions

Figure 7 Nek7 Knockdown Decreases PV+ Interneuron Outputs (A) Schematic of the experimental design. Cre-dependent virus expressing shRNA and the fluorescent marker mCherry were injected in E15.5 (left) and P3 (right) Lhx6Cre mice in vivo. Boutons co-stained with SYT2 and mCherry were quantified either inside PV+ arbors or onto NeuN+ somata. (B and C) Confocal Z projections and Imaris 3D reconstructions showing control shRNA (B) and Nek7 shRNA (C) infected PV+ cells expressing mCherry (red) in axons containing SYT2+ boutons (blue). (D) SYT2+ bouton density per unit area of neurite comparing control shRNA (n = 24 PV+ neurons from four mice) and Nek7 shRNA (n = 22 PV+ neurons from six mice). Student’s t test. (E–G) Confocal images and 3D Imaris reconstructions showing mCherry+ (red), SYT2+ (green) synaptic boutons from infected PV+ cells expressing control shRNA (E), Nek7 shRNA (F), and Nek7 shRNA with mNek7 for rescue (G) contacting pyramidal cells NeuN+ (blue). SYT2+ mCherry+ (filled arrowheads, yellow spheres), SYT2+ mCherry− (open arrowheads, green spheres). (H) Percentage of SYT2+ mCherry+ somatic boutons contacting the pyramidal cells normalized to the percentage of PV+ cells infected in the area, comparing shRNA (n = 182 PV+ neurons from seven mice), Nek7 shRNA (n = 200 pyramidal cells from six mice), and mNek7 (n = 109 pyramidal cells from four mice). Kruskal-Wallis test, pairwise comparisons. ∗p < 0.05 and ∗∗p < 0.01; n.s., not significant. Data are represented as mean ± SEM. Scale bars represent 2 μm (B and C) and 5 μm (E–G). See also Figure S6. Cell Reports 2018 24, 1231-1242DOI: (10.1016/j.celrep.2018.06.115) Copyright © 2018 Terms and Conditions